3-Aryl-2-Quinolone Derivatives: Synthesis and Characterization of In

Indeed, the concentration values decreasing the growth of the 12 cell lines by at least 50% (IC50 index) were always higher than 10-5 M. We then made ...
0 downloads 7 Views 232KB Size
J. Med. Chem. 2002, 45, 2543-2555

2543

3-Aryl-2-Quinolone Derivatives: Synthesis and Characterization of In Vitro and In Vivo Antitumor Effects with Emphasis on a New Therapeutical Target Connected with Cell Migration Benoıˆt Joseph,*,† Francis Darro,‡ Aure´lie Be´hard,† Brigitte Lesur,‡ Franc¸ oise Collignon,‡ Christine Decaestecker,§ Armand Frydman,‡ Ge´rald Guillaumet,† and Robert Kiss§ Institut de Chimie Organique et Analytique UMR-CNRS 6005, Universite´ d’Orle´ ans, BP 6759, 45067 Orle´ ans Cedex 2, France, Laboratoire L. Lafon, 19, Avenue du Professeur Cadiot, BP 22, 94701 Maisons-Alfort Cedex, France, and Laboratoire d’Histopathologie, Faculte´ de Me´ decine, Universite´ Libre de Bruxelles, CP620, Route de Lennik 808, B-1070 Bruxelles, Belgique Received July 6, 2001

Among 25 3-aryl-2-quinolone derivatives synthesized, the antitumor activity of some of them was characterized both in vitro and in vivo. In this series, no compound appeared to be cytotoxic in vitro, as was known by the colorimetric MTT assay carried out on 12 distinct human cancer cell lines obtained from the American Type Culture Collection. Indeed, the concentration values decreasing the growth of the 12 cell lines by at least 50% (IC50 index) were always higher than 10-5 M. We then made use of a computer-assisted phase-contrast videomicroscopy system to quantitatively determine in vitro the level of migration of living MCF-7 human breast cancer cells. For example, at 10-7 M, compounds 7, 13, 16, and 28 markedly decreased the migration level of these MCF-7 human breast cancer cells. The in vivo determination of the maximum tolerated dose showed that all compounds tested were definitively nontoxic. When the nontoxic, antimigratory compound 16 was combined with either doxorubicin or etoposide, two cytotoxic compounds routinely used in the clinic, this led to additive in vivo benefits from this treatment (as compared to individual administrations of the drugs) when the MXT mouse mammary adenocarcinoma was used. Thus, nontoxic antimigratory compounds, including the 2-quinolone derivatives synthesized here, can actually improve the efficiency of antitumor treatment when combined with conventional cytotoxic agents. Introduction From a schematic point of view, the development of malignancy in a given tissue occurs as follows. Some genomic mutations perturb cell cycle kinetics by increasing cell proliferation or decreasing cell death (or both), and these features lead to unrestrained growth of the genomically transformed cell population.1 Then, some cells from this transformed cell population switch to the angiogenic phenotype, and this enables them to recruit endothelial cells from the healthy tissue, leading to a neoangiogenic process enabling the sustained growth of the developing neoplastic tissue.2 Then, some cells migrate from the tumor bulk and colonize new tissues (the metastatic process), using blood or lymphatic vessels as major routes of migration.3 The first chemotherapeutic agents used against cancer in clinics were drugs that target processes including nucleoside synthesis, strand replication, etc., i.e., processes controlling DNA replication and therefore cell proliferation.4 The discovery of programmed cell death (apoptosis) by Kerr and colleagues in the early 1970s5 was evidence that rapidly growing tumors are not always tumors exhibiting high levels of cell proliferation but often low levels of cell death as compared to the normal cell * To whom correspondence should be addressed. Tel.: (33)238 49 45 85. Fax: (33)238 41 72 81. E-mail: [email protected]. † Universite ´ d’Orle´ans. ‡ Laboratoire L. Lafon. § Universite ´ Libre de Bruxelles.

population from which these tumor cells issue.6 These findings thus led to the development of anticancer drugs activating the cell death of tumor cells rather than inhibiting their proliferation.7 Most of these agents target topoisomerases, which play crucial roles in DNA duplication.8 They include, for example, doxorubicin (DOX) and etoposide (ETO) as inhibitors of topoisomerase II and irinotecan and topotecan as inhibitors of topoisomerase I. Thus, most of the agents used today in hospitals to treat cancer patients are drugs that more or less directly target the cell kinetics (i.e., proliferation vs death) of the cancer to be combatted. Great hopes are placed in complementary strategies including antiangiogenic,9 immunotherapeutic,10 and gene therapy11 approaches. The fact nevertheless remains that the great majority of the drugs used in the standard treatment of cancer are toxic to highly toxic, and this limits their clinical use to a relatively low number of administrations per patient. In addition, several of these compounds must be combined into polychemotherapeutic regimens in order to have any effect against cancer. By way of evidence, such anticancer drug combinations increase the toxicity of the treatment and once again limit the number of administrations that can be applied per patient. In addition, a hallmark of malignancy, which is as important as cell kinetics, is the migration of cancer cells escaping from the tumor bulk that first invade neighboring tissue and then establish metastases. Antitubulin compounds, one major class of antican-

10.1021/jm010978m CCC: $22.00 © 2002 American Chemical Society Published on Web 05/11/2002

2544

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

cer drugs, already target tumor cell migration or at least partly.12 While effective against human cancers, these compoundssincluding, for example, vincristine, vinorelbine, and paclitaxelsare nevertheless also toxic. It has been known for many years that flavonols, as quercetin, are polyphenols, which act as free radical scavangers and could be significant anticarcinogenic substances (through food intake) due to not yet welldefined biological mechanisms.13 Such flavonoids inhibit proliferation of the MCF-7 human breast cancer cell line.14 Yoshida et al. have suggested that antiproliferative effects of quercetin were due to the specific arrest of the G1 phase of the cell cycle.15 However, the results of epidemiological studies reported so far do not show a clear protective effect of flavonol intake (through food and tea consumption) on cancer risk but a protective effect on selected cancers in a specific population cannot be ruled out. Because of that, hypotheses are proposed that a protective role of quercetin (from onions) in explaining the lower risk of stomach cancer might be due to interaction with early processes in tumor development (i.e., angiogenesis, cell migration, etc.). For this reason, we have considered the 2-quinolone template as a possible source of chemical modulation when looking at the benzopyrane-4-one ring. Moreover, variations on the 3-aryl moiety were designed with the variety of compounds known in the flavonoid/isoflavonoid families in mind. We have prepared 25 3-aryl-2-quinolone derivatives A with the aim of developing nontoxic antimigratory compounds that could complement the anticancer action of conventional drugs already used in hospitals. Some rationale as to why 3-aryl-2-quinolones can be antimigratory agents is given in the discussion.

Joseph et al.

Scheme 1a

a Reagents and conditions: (a) Toluene, room temperature, 1 h. (b) (i) POCl3, DMF, -30 °C to 75 °C; (ii) AcOH/H2O, reflux 3 h.

Scheme 2a

a

Reagents and conditions: (a) PPSE, 110 °C, 2 h.

Scheme 3a

Chemistry The syntheses of 3-aryl-2-quinolone derivatives have been accomplished as described in Schemes 1-6. According to the literature method,16,17 the key 3-aryl-2quinolones 2a-g were synthesized. As described in Scheme 1, substituted anilines were acylated with phenylacetyl chloride, 4-methoxyphenylacetyl chloride, or 4-benzyloxyphenylacetyl chloride to give the corresponding amides 1a-g,18 which were then treated with phosphorus oxychloride and N,N-dimethylformamide (DMF) followed by an acid hydrolysis to give 3-aryl-2quinolones 2a-g. Compounds 2c,e were also prepared through a one pot route (Scheme 2). Thus, the condensation of 3,5-dimethoxyaniline and ethyl 2-(phenyl or 4-methoxyphenyl)-2-formyl acetate19 was carried out in the presence of polyphosphoric acid trimethylsilyl ester (PPSE)20 at 110 °C for 2 h to give the desired compounds 2c,e, respectively, in 16 and 32% yield. In an independent but closed work published in 1997, Bisagni et al. observed the same results by thermal cyclization of 3,5dimethoxyaniline and ethyl 2-(4-methoxyphenyl)-2formyl acetate.21

a Reagents and conditions: (a) NaH, RX, DMF, 0 °C and then 90 °C. (b) H2, Pd-C 10%, 1,4-dioxane, room temperature, 4 h.

Alkylation of quinolone 2e led to the preparation of target compounds 3-10 and O-alkylated derivatives 3a-10a (Scheme 3). In all cases, the N-alkylated derivative was obtained as the major product. The structure of both series of compounds was unambiguously confirmed by 13C nuclear magnetic resonance (NMR) (i.e., for 5, δ 45.3 (NCH2) and for 5a, δ 63.0 (OCH2)) and by analysis of the literature published on this topic.22 Catalytic hydrogenolysis of 10 led to the acid 11. Compounds 12-14 bearing a functionalized ethylenic chain were obtained via a Michael addition with

3-Aryl-2-Quinolone Derivatives

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

Scheme 4a

Scheme 5a

2545

a Reagents and conditions: (a) 48% HBr, AcOH reflux, 3 days. (b) 48% HBr, AcOH, reflux, 5 h. (c) Lawesson’s reagent, toluene, reflux, 18 h. (d) (Ac)2O, pyridine, room temperature, 18 h. a Reagents and conditions: (a) CH dCHR, Triton B, DMF, room 2 temperature, 18 h. (b) H2, Pd/C 10%, 1,4-dioxane, room temperature, 48 h. (c) Bu3SnN3, toluene, 105 °C, 65 h. (d) DCC, HOBt, (Et)2NH, DMF, room temperature, 24 h. (e) PPA, P2O5, 110 °C, 45 min.

various acrylates (Scheme 4). Catalytic hydrogenolysis of 14 led to the acid 15. Compound 13 was transformed into the 5-tetrazolylethyl derivative 16 by using tributyltin azide (Scheme 4). The dicyclohexylcarbodiimide/ 1-hydroxybenzotriazole coupling method was applied to 15 to give compound 17 in good yield (Scheme 4). Similarly, polyphosphoric acid cyclization of 15 led to the preparation of tricyclic target compound 18 (Scheme 4). The O-demethylation of 2e and 3 was performed with 48% HBr in acetic acid to give, respectively, 19 and 20 in 3 days or 21 and 22 in 5 h (Scheme 5). Compounds 2e and 3 were converted into the corresponding 2-thioquinolones 23 and 24 by treatment with Lawesson’s reagent in toluene (Scheme 5). Compounds 25 and 26 were generated by acetic anhydride-pyridine acetylation of 20 and 22 in 60% yield (Scheme 5). Treatment of 24 with iodomethane in tetrahydrofuran (THF) afforded the iodide salt 27 in 79% yield. Reaction of 27 with phenylhydrazine or 2-pyridinylhydrazine in refluxing ethanol gave hydrazones 28 and 29 in fair yields (Scheme 6). The yields and/or analytical data of compounds 1-26, 28, and 29 are summarized in Tables 1-3. Pharmacological Results and Discussion The absence of in vitro cytotoxicity of 3-aryl-2-quinolone derivatives so prepared was demonstrated by means of the MTT colorimetric assay23 on 12 distinct human cancer cell lines. The results show that when compared to the control value when assayed at 10-5 M, 19 of the 25 2-quinolone derivatives synthesized significantly decreased the mean growth value of the 12 human cancer cell lines under study, while the remain-

Scheme 6a

a Reagents and conditions: (a) ICH , THF, room temperature, 3 12 h. (b) NH2NHPh or NH2NH-2-Pyr, EtOH, sealed tube, 90 °C.

ing six compounds 2e, 7, 15, 19, 20, and 23 were without any actual effect at this 10-5 M dose. At the 10-6 M dose, only few compounds significantly decreased the growth levels of human cancer cell populations. At the 10-7 M dose, only two compounds (17 and 21) showed marginal cytotoxic effects. The determination of the maximum tolerated dose (MTD) index in vivo revealed that all quinolones prepared were found nontoxic at 160 mg/ kg. The effects of these quinolone derivatives were then characterized in vitro on MCF-7 human breast cancer cell motility by means of computer-assisted phasecontrast videomicroscopy.24 The migration features of cancer cells involve two distinct but complementary processes, i.e., motility and invasion. Motility refers to the capacity of cells to move (including polymerization/ depolymerization dynamics of the actin skeleton and integrin moving in adhesion complexes), while invasion refers to the capacity of cells to modulate their surrounding environment by proteolytic activity. The system that we set up thus enabled the motility level of living cells to be quantitatively determined in vitro. However, we showed with respect to human glioma cancer cells that their in vitro motility characteristics,

2546

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

Joseph et al.

Table 1. Structures, Yields, and Physical Data of Compounds 1 and 2

compd

R

R1

R2

R3

R4

yield (%)

mp (°C)

formula

anal

1a 1b 1c 1d 1e 1f 1g 2a 2b 2c 2d 2e 2f 2g

H H H OCH3 OCH3 OCH3 OBn H H H OCH3 OCH3 OCH3 OBn

H H OCH3 H OCH3 OCH3 OCH3 H H OCH3 H OCH3 OCH3 OCH3

H OCH3 H H H H H H OCH3 H H H H H

H H OCH3 H OCH3 H OCH3 H H OCH3 H OCH3 H OCH3

OCH3 H H OCH3 H OCH3 H OCH3 H H OCH3 H OCH3 H

92 89 87 91 82 93 81 10 28 16Meth.A,B 10 22Meth.A, 32Meth.B 45 20

80-81a,b 118-119c,d 109-111a 47-48a 135-137a,e 89-90a 122-123c 188-189f 243-244f 257-258f,g 148-149f 254-255f,h 186-187f 234-235f

C15H15NO2 C15H15NO2 C16H17NO3 C16H17NO3 C17H19NO4 C17H19NO4 C23H23NO4 C16H13NO2 C16H13NO2 C17H15NO3 C17H15NO3 C18H17NO4 C18H17NO4 C24H21NO4

C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N

a Toluene (recrystallization solvent). b Literature18 85 °C. c EtOAc/PE (recrystallization solvent). d Literature18 123 °C. e Literature21 150 °C. f EtOAc (recrystallization solvent). g Literature17 243 °C. h Literature21 260 °C; Meth. A, Meth. B, see experimental section.

Table 2. Structures and Physical Data of Compounds 3-18

compd 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17

R5 CH3 CH2COOCH2CH3 CH2CON(CH2CH3)2 CH2CH2N(CH3)2 CH2CH2CH2N(CH3)2 CH2CN CH2CH2CH2CN CH2COOBn CH2COOH CH2CH2COOCH3 CH2CH2CN CH2CH2COOBn CH2CH2COOH 5-tetrazolylethyl CH2CH2CON(CH2CH3)2

18

a

Recrystallization solvent: EtOAc/PE. b EtOAc. c Et2O.

d

mp (°C)

formula

anal

125-126a 160-161a 178-179b 113-114c 94-95c 208-209b 157-158b 199-200c 179-180c 100-101b 156-157a 124-125d 194-195c 234-235c 137-138b

C19H19NO4 C22H23NO6 C24H28N2O5 C22H26N2O4 C23H28N2O4 C20H18N2O4 C22H22N2O4 C27H25NO6 C20H19NO6 C22H23NO6 C21H20N2O4 C28H27NO6 C21H21NO6 C21H21N5O4 C25H30N2O5

C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N

240-241e

C21H19NO5

C, H, N

Et2O/PE. e CH2Cl2/PE.

as determined by the system that we set up, correlate with their in vivo invasive capacities.24,25 The fact that antimigratory effects observed in vitro for a given compound can relate to antimetastatic potential is already demonstrated for inhibitors of hepatocyte growth factor receptor.26,27 Preliminary data indicated that this first series of quinolone derivatives exhibits interacting effects at the cell migration level (data not shown) with inhibition of migration effects seen for various chemical types of R5 or X radicals: for example, compounds 7 (X ) O, R5 ) -(CH2)3-N(CH3)2), 13 (X ) O, R5 ) -(CH2)2-CN), 16 (X ) O, R5 ) 5-tetrazoylethyl), and 28 (X ) NNHPh,

R5 ) -CH3). Figure 1 illustrates the antimigratory effects obtained with respect to compounds 13 and 16. Both compounds induce a significant decrease in MCF-7 human breast cancer cell migration, with more marked and dose-dependent effects for compound 16 than 13. Compound 16 had a significant antimigratory effect on the MCF-7 cells at the definitively noncytotoxic 10-7 M dose. At the lowest dose tested for compound 16, i.e., 10-8 M, a slight stimulation of cell migration was observed (Figure 1B). 3-Aryl-2-quinolones are reported to be inhibitors of arachidonic acid-induced platelet agregation28 and also related to 3-arylpyridopyrimidines that are reported to

3-Aryl-2-Quinolone Derivatives

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

2547

Table 3. Structures and Physical Data of Compounds 19-26, 28, and 29

compd

R

R1

R3

R5

X

mp (°C)

formula

anal

19 20 21 22 23 24 25 26 28 29

H H H H CH3 CH3 Ac Ac CH3 CH3

H H CH3 CH3 CH3 CH3 Ac CH3 CH3 CH3

H H CH3 CH3 CH3 CH3 Ac CH3 CH3 CH3

H CH3 H CH3 H CH3 CH3 CH3 CH3 CH3

O O O O S S O O NNHPh NNH-2-Pyr

>280a,b >280b 275-276b 204-205b 229-230c 176-177d 206-207b 148-149d 148-149e 140-141e

C15H11NO4 C16H13NO4 C17H15NO4 C18H17NO4 C18H17NO3S C19H19NO3S C22H19NO7 C20H19NO5 C25H25N3O3 C24H24N4O3

C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N C, H, N

a

Literature21 >280 °C. b Recrystallization solvent: EtOAc. c Et2O.

Figure 1. Characterization of the antimigratory effects induced by compounds 13 (A) and 16 (B) on the migration level of the human MCF-7 breast cancer cells. The migration level was quantitatively determined by means of computer-assisted phase-contrast videomicroscopy on several hundreds of cells per experimental condition. The data are reported as mean ( SEM.

be both kinase receptor inhibitors29 and angiogenesis inhibitors.30 These data already published in the literature can have some relevance with our data and can therefore suggest potential 3-aryl-2-quinolone derivative-mediated effects on distinct types of kinase receptors that in turn will significantly decrease the motility levels of human breast cancer cells. Compound 16 was then combined with either DOX or ETO (which are two clinically active anticancer drugs) in order to investigate in vivo the MXT mouse mammary adenocarcinoma,31,32 which closely mimics

d

EtOAc/PE. e MeOH.

human breast cancer,33,34 and whether any benefit could be obtained from additive treatment with such combinations. We chose the MXT mouse mammary adenocarcinoma model originating from ductal structures33,34 for the following reasons. The first concerns the fact that up to 1986 leukemia models were the NCI’s preferred models for testing new investigational agents in vivo, but these models were then abandoned because they are too chemosensitive and consequently fail to reflect clinical reality.35 We thus preferred to make use of a solid tumor. Second, the MXT mouse mammary cancer was selected because more than 80% of female breast cancers are invasive intraduct carcinomas, i.e., not otherwise specified breast cancers,33 while most of the experimental murine mammary tumors originate from the glandular acini.33 The biological characteristics of mammary tumors from ductal as opposed to acinar structures differ markedly.36 Figure 2 illustrates the effects obtained with ETO (A), DOX (B), and compound 16 (C). The data indicate that ETO significantly (P < 0.05 to P < 0.001 as compared to control at the different days postgraft) decreased the MXT tumor growth when tested at 20 (MTD/4) or 10 (MTD/8) mg/kg (Figure 2A). At 40 mg/kg (MTD/2), ETO showed itself to be highly toxic, while at 5 mg/kg (MTD/16), it became ineffective (Figure 2A). We chose the 10 mg/kg ETO dose for a combined treatment with compound 16 (Figure 3A). The same experimental approach was used with respect to DOX, which was tested at MTD/2 (5 mg/kg), MTD/4 (2.5 mg/kg), MTD/8 (1.25 mg/kg), and MTD/16 (0.63 mg/kg). The MTD/2 treatment appeared highly toxic, while the MTD/16 treatment was ineffective (Figure 2B). The MTD/4 treatment (P < 0.05 to P < 0.01) was more effective than the MTD/8 treatment (P < 0.05 to P < 0.001) and was chosen for the combined treatment with compound 16 (Figure 3B). This latter was assayed at MTD/2 (80 mg/kg), MTD/4 (40 mg/kg), and MTD/8 (20 mg/kg). Only 80 mg/kg, the highest dose tested, showed slight, but nevertheless significant (P < 0.05), antitumor effects when compared to the control (Figure 2C). The 40 mg/kg dose (which exhibited no significant effects per se; Figure 2C) was the one chosen for combined treatment with either ETO (Figure 3A) or DOX (Figure 3B). The data obtained revealed that real additive anticancer influences were observed for both compound 16/

2548 Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

Figure 2. Characterization of the anticancer influence of ETO (A), DOX (B), and 16 (C) on the growth of the MXT mouse mammary adenocarcinoma. ETO was tested at 40 (MTD/2; O), 20 (MTD/4; ]), 10 (MTD/8; 0), and 5 (MTD/16; ×) mg/kg. DOX was tested at 5 (MTD/2; O), 2.5 (MTD/4; ]), 1.25 (MTD/8; 0), and 0.63 (MTD/16; ×) mg/kg. Compound 16 was tested at 80 (MTD/2; O), 40 (MTD/4; ]), and 20 (MTD/8; 0). The control conditions are symbolized by black dots. ETO and DOX were administered i.p. (0.2 mL) nine times according to the experimental schedule depicted by the black arrows in the upper left-hand part of panels A and B. Derivative 16 was administered 20 times (see the black arrows in the upper left-hand part of panel C). There were nine mice per experimental group. The P levels of statistical significance were determined by means of the nonparametric Mann-Whitney test. The results are depicted as mean values (the symbols) ( their SEM (the bars).

ETO (Figure 3A) and compound 16/DOX (Figure 3B) combinations, as was evidenced by the increases in the P levels of statistical significance depicted in Figure 3A,B. Indeed, a slight, but nevertheless significant, effect (P ) 0.04) was obtained when using 10 mg/kg of ETO with 40 mg/kg of compound 16, with an actual increase observed in the survival periods of the MXT mammary cancer-bearing mice, which increased by 38% (P ) 0.01) as compared to the control (Figure 3A). The

Joseph et al.

Figure 3. Characterization of the anticancer influence exercised by ETO combined with 16 (× in A) and by DOX combined with 16 (× in B) on the growth of the MXT mouse mammary adenocarcinoma. ETO was assayed at 10 mg/kg, i.e., at MTD/8 (0 in A), DOX at MTD/4 (2.5 mg/kg; 4 in B), and 16 at MTD/4 (40 mg/kg; O in A and B). The control conditions are symbolized by black dots. There were nine mice per experimental group. The P levels of statistical significance were determined by means of the nonparametric Mann-Whitney test. The results are depicted as mean values (the symbols) ( their SEM (the bars). The P values in A and B were calculated at the end of the experiments (see the vertical dotted line) when at least four mice survived in each experimental group.

combination of compound 16 (40 mg/kg) and DOX (2.5 mg/kg) also significantly (P ) 0.008) increased the antitumor effects observed with each compound tested individually (Figure 3B). The P values in Figure 3A,B were calculated at the end of the experiments (see the vertical dotted line) when at least four mice survived in each experimental group. These additive effects observed in terms of antitumor activities between compound 16 and DOX or ETO can result from a combined antimigratory effect brought by compound 16 on the tumor cells and an antiproliferative effect brought by ETO or DOX on the remaining tumor cells. In conclusion, the data from the present study clearly indicate that an improvement in treatment was obtained in the case of the MXT mouse mammary tumor with a combination of the nontoxic antimigratory compound 16 (taken up as a representative of the group of study drugs) and the conventional cytotoxic drugs used routinely in hospitals, i.e., either ETO or DOX, which are two topoisomerase II inhibitors. The mechanism of action of compound 16 and congeners still remains to be elucidated. Further combinations with various other

3-Aryl-2-Quinolone Derivatives

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

anticancer drugs are currently being investigated against several other cancer cell lines.

to the procedure described for 1a. IR (KBr): ν 3292, 1658, 1615 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.66 (s, 2H, CH2), 3.74 (s, 6H, CH3), 3.82 (s, 3H, CH3), 6.20 (t, 1H, J ) 2.2 Hz, HAr), 6.62-6.66 (m, 2H, HAr), 6.95 (d, 2H, J ) 7.5 Hz, HAr), 6.97 (br s, 1H, NH), 7.23 (d, 2H, J ) 7.5 Hz, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.0, 55.3, 55.4 (2), 96.7, 97.9 (2), 114.4 (2), 126.2, 130.7 (2), 139.4, 159.0, 161.0 (2), 169.5. MS: m/z 302 (M+ + 1). N-(2,5-Dimethoxyphenyl)-2-(4-methoxyphenyl)acetamide (1f). Starting from 2,5-dimethoxyaniline and 4-methoxyphenylacetyl chloride, compound 1f was obtained according to the procedure described for 1a. IR (KBr): ν 3292, 1659, 1614 cm-1. 1H NMR (250 MHz, CDCl3): δ 3,67 (s, 3H, CH3), 3.68 (s, 2H, CH2), 3.75 (s, 3H, CH3), 3.81 (s, 3H, CH3), 6.53 (dd, 1H, J ) 3.0, 9.0 Hz, HAr), 6.71 (d, 1H, J ) 9.0 Hz, HAr), 6.92 (d, 2H, J ) 8.8 Hz, HAr), 7.25 (d, 2H, J ) 8.8 Hz, HAr), 7.82 (br s, 1H, NH), 8.80 (d, 1H, J ) 3.0 Hz, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.2, 55.3, 55.7, 56.3, 105.5, 108.6, 110.9, 114.4 (2), 126.4, 128.3, 130.6 (2), 142.0, 153.9, 158.9, 169.3. MS: m/z 302 (M+ + 1). N-(3,5-Dimethoxyphenyl)-2-(4-benzyloxyphenyl)acetamide (1g). Starting from 3,5-dimethoxyaniline and 4-benzyloxyphenylacetyl chloride, compound 1g was obtained following a procedure as described for 1a. The crude residue was purified by column chromatography (eluent ethyl acetate/ petroleum ether 6:4) to afford 1g. IR (KBr): ν 3291, 1659, 1610 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.66 (s, 2H, CH2), 3.74 (s, 3H, CH3), 3.75 (s, 3H, CH3), 5.08 (s, 2H, CH2), 6.21 (t, 1H, J ) 2.2 Hz, HAr), 6.66 (d, 2H, J ) 2.2 Hz, HAr), 7.00 (d, 2H, J ) 8.8 Hz, HAr), 7.08 (br s, 1H, NH), 7.24 (d, 2H, J ) 8.8 Hz, HAr), 7.33-7.46 (m, 5H, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.2, 55.5 (2), 70.2, 96.9, 98.0 (2), 115.7 (2), 126.6, 127.6 (2), 128.2, 128.8 (2), 130.8 (2), 136.9, 139.6, 158.4, 161.1 (2), 169.7. MS: m/z 378 (M+ + 1). General Procedure for Preparation of Compound 2. 8-Methoxy-3-phenyl-1,2-dihydro-2-quinolinone (2a). To a solution of POCl3 (1.75 mL, 19 mmol) at -30 °C was dropwise added anhydrous DMF (0.31 mL, 4.0 mmol). The solution was stirred for 15 min at -30 °C, and then, compound 1a (2.7 mmol) was dropwise added. Under vigorous stirring, the reaction mixture was allowed to reach room temperature and was then heated at 75 °C for 1.5 h. The solution was poured into ice, neutralized with 30% aqueous NH4OH, and extracted with CH2Cl2. The organic layer was dried over MgSO4 and then evaporated in vacuo. The crude residue obtained was dissolved in glacial acetic acid (4.75 mL) and H2O (0.15 mL). The final solution was stirred at reflux for 3 h. After it was cooled, the solvent was evaporated in vacuo. The crude residue was diluted in H2O, neutralized with 25% NaOH, and finally extracted with CH2Cl2. The organic layer was dried over MgSO4 and then evaporated in vacuo. The crude residue was crystallized from EtOAc. Crystals was filtered and washed twice with EtOAc to give 2a. IR (KBr): ν 1646, 1607 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.92 (s, 3H, CH3), 7.13-7.16 (m, 2H, HAr), 7.30-7.46 (m, 4H, HAr), 7.74-7.77 (m, 2H, HAr), 8.09 (s, 1H, dCH), 10.98 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 56.5, 111.3, 120.3, 120.4, 128.3 (2), 128.4, 128.7, 129.2 (2), 132.6, 136.7, 138.2, 145.9, 161.1. MS: m/z 252 (M+ + 1). 6-Methoxy-3-phenyl-1,2-dihydro-2-quinolinone (2b). According to the procedure described for 2a, the 2-quinolone 2b was prepared from 1b. IR (KBr): ν 1645, 1618 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.80 (s, 3H, CH3), 7.16 (dd, 1H, J ) 2.5, 8.9 Hz, HAr), 7.28 (d, 1H, J ) 8.9 Hz, HAr), 7.29 (d, 1H, J ) 2.5 Hz, HAr), 7.34-7.47 (m, 3H, HAr), 7.76 (d, 2H, J ) 6.8 Hz, HAr), 8.06 (s, 1H, dCH), 11.85 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 55.4, 109.4, 116.0, 119.5, 120.1, 127.8, 127.9 (2), 128.7 (2), 131.9, 132.9, 136.4, 137.2, 154.2, 160.6. MS: m/z 252 (M+ + 1). 5,7-Dimethoxy-3-phenyl-1,2-dihydro-2-quinolinone(2c).17 Method A. According to the procedure described for 2a, the 2-quinolone 2c was prepared from 1c. Method B. To a solution of 2-phenyl-2-formyl acetate (7.37 g, 38.4 mmol) and 3,5dimethoxyaniline (5.0 g, 32 mmol) stirred at room temperature

Experimental Section Chemistry. Melting points were determined using a Bu¨chi capillary instrument and are uncorrected. IR spectra were recorded on a Perkin-Elmer FTIR Paragon 1000 spectrophotometer. NMR spectra were recorded at 300 K on a Bruker Avance DPX 250 spectrometer. Chemical shifts are expressed in parts per million relative to tetramethylsilane (TMS), and spin multiplicities are given as s (singlet), br s (broad singlet), d (doublet), dd (double doublet), t (triplet), and m (multiplet). Mass spectra were recorded on a Perkin-Elmer SCIEX API 300 instrument using ionspray methodology. Elemental compositions of the compounds agreed to within 0.4% of the calculated value. Thin-layer chromatography (TLC) was run on precoated silica gel plates (Merck 60F254), and spots were visualized with a UV light at 254 nm. Column chromatography was carried out using Merck silica gel (230-400 mesh). Most chemicals and solvents were analytical grade and used without further purification. Ethyl 2-(phenyl or 4-methoxyphenyl)-2formyl acetates were prepared according to ref 19. Commercial reagents were purchased from Acros Company. General Procedure for Preparation of Compound 1. N-(2-Methoxyphenyl)-2-phenylacetamide (1a).18 To a stirred solution of o-anisidine (1.37 mL, 10.0 mmol) in toluene (14 mL) at 0 °C was added dropwise a solution of phenylacetyl chloride (1.62 mL, 10.0 mmol) in toluene (5 mL). The reaction mixture was stirred at room temperature for 1 h and then treated with a saturated NaHCO3 solution. The biphasic solution was vigorously stirred for 30 min, then decanted, and finally separated. The collected aqueous phase was extracted with EtOAc (2 × 10 mL). The combined organic layer was dried over MgSO4 and evaporated. The crude oily residue was crystallized from toluene to yield 1a. IR (KBr): ν 3287, 1652, 1598 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.72 (s, 3H, CH3), 3.76 (s, 2H, CH2), 6.81 (dd, 1H, J ) 1.8, 8.0 Hz, HAr), 6.917.05 (m, 2H, HAr), 7.21-7.37 (m, 5H, HAr), 7.80 (br s, 1H, NH), 8.35 (dd, 1H, J ) 1.8, 8.0 Hz, HAr). 13C NMR (62.90 MHz, CDCl3): δ 45.2, 55.7, 109.9, 119.5, 121.1, 123.7, 127.4, 127.6, 129.0 (2), 129.6 (2), 134.6, 147.8, 168.8. MS: m/z 242 (M+ + 1). N-(4-Methoxyphenyl)-2-phenylacetamide (1b).18 Starting from p-anisidine and phenylacetyl chloride, compound 1b was obtained according to the procedure described for 1a. IR (KBr): ν 3290, 1650, 1603 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.72 (s, 2H, CH2), 3.77 (s, 3H, CH3), 6.81 (d, 2H, J ) 9.0 Hz, HAr), 7.00 (br s, 1H, NH), 7.28-7.43 (m, 7H, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.8, 55.6, 114.2 (2), 121.9 (2), 127.7, 129.3 (2), 129.7 (2), 130.8, 134.7, 156.7, 169.1. MS: m/z 242 (M+ + 1). N-(3,5-Dimethoxyphenyl)-2-phenylacetamide (1c). Starting from 3,5-dimethoxyaniline and phenylacetyl chloride, compound 1c was obtained according to the procedure described for 1a. IR (KBr): ν 3286, 1657, 1616 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.70 (s, 2H, CH2), 3.74 (s, 6H, CH3), 6.21 (t, 1H, J ) 2.2 Hz, HAr), 6.66 (d, 2H, J ) 2.2 Hz, HAr), 7.09 (br s, 1H, NH), 7.30-7.40 (m, 5H, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.9, 55.4 (2), 96.8, 97.9 (2), 127.7, 129.2 (2), 129.5 (2), 134.3, 139.4, 161.0 (2), 169.1. MS: m/z 272 (M+ + 1). N-(2-Methoxyphenyl)-2-(4-methoxyphenyl)acetamide (1d). Starting from o-anisidine and 4-methoxyphenylacetyl chloride, compound 1d was obtained according to the procedure described for 1a. IR (KBr): ν 3375, 1667, 1597 cm-1. 1H NMR (250 MHz, CDCl3): δ 3,68 (s, 2H, CH2), 3.72 (s, 3H, CH3), 3.82 (s, 3H, CH3), 6.79 (dd 1H, J ) 1.5, 8.0 Hz, HAr), 6.897.03 (m, 4H, HAr), 7.25 (d, 2H, J ) 8.8 Hz, HAr), 7.79 (br s, 1H, NH), 8.33 (dd, 1H, J ) 1.5, 8.0 Hz, HAr). 13C NMR (62.90 MHz, CDCl3): δ 44.2, 55.3, 55.7, 109.9, 114.4 (2), 119.5, 121.0, 123.6, 126.6, 127.6, 130.7 (2), 147.8, 158.9, 169.3. MS: m/z 272 (M+ + 1). N-(3,5-Methoxyphenyl)-2-(4-methoxyphenyl)acetamide (1e).21 Starting from 3,5-dimethoxyaniline and 4-methoxyphenylacetyl chloride, compound 1e was obtained according

2549

2550

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

Joseph et al.

for 30 min was added a freshly prepared solution of PPSE (15.0 g of phosphorus pentoxide and 36 mL of hexamethyldisiloxane in 1,2-dichloroethane stirred at 90 °C until complete dissolution of P2O5 and then evaporation of solvent). The reaction mixture was stirred at 100 °C for 1 h. After the mixture was cooled, CH2Cl2 (1000 mL) and H2O (500 mL) were added to the solution. The biphasic solution was vigorously stirred overnight, then decanted, and finally separated. The collected aqueous phase was extracted with CH2Cl2 (2 × 100 mL). The combined organic layer was dried over MgSO4 and evaporated. The crude oily residue was crystallized from EtOAc to give 2c. IR (KBr): ν 1668, 1631 cm-1. 1H NMR (250 MHz, DMSOd6): δ 3.81 (s, 3H, OCH3), 3.89 (s, 3H, OCH3), 6.36 (d, 1H, J ) 1.8 Hz, HAr), 6.45 (d, 1H, J ) 1.8 Hz, HAr), 7.28-7.42 (m, 3H, HAr), 7.69 (d, 2H, J ) 7.0 Hz, HAr), 8.00 (s, 1H, dCH), 11.81 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 55.5, 56.0, 90.0, 93.1, 104.5, 126.9, 127.3, 127.9 (2), 128.4 (2), 131.4, 136.7, 140.8, 156.8, 161.4, 162.2. MS: m/z 282 (M+ + 1). 8-Methoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinone (2d). According to the procedure described for 2a, the 2-quinolone 2d was prepared from 1d. IR (KBr): ν 1652, 1625 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.79 (s, 3H, CH3), 3.90 (s, 3H, CH3), 6.99 (d, 2H, J ) 8.8 Hz, HAr), 7.09-7.15 (m, 2H, HAr), 7.29 (dd, 1H, J ) 3.2, 6.0 Hz, HAr), 7.74 (d, 2H, J ) 8.8 Hz, HAr), 8.03 (s, 1H, dCH), 10.90 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 55.2, 56.0, 110.5, 113.4 (2), 119.6, 120.0, 121.8, 127.9, 128.5, 129.9 (2), 131.6, 136.4, 145.3, 159.1, 160.7. MS: m/z 282 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinone (2e).21 According to the procedures described for 2c, the 2-quinolone 2e was prepared from 1e (method A) or 2-(4methoxyphenyl)-2-formyl acetate and 3,5-dimethoxyaniline (method B). IR (KBr): ν 1664, 1628 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.78 (s, 3H, CH3), 3.81 (s, 3H, CH3), 3.89 (s, 3H, CH3), 6.35 (d, 1H, J ) 2.0 Hz, HAr), 6.45 (d, 1H, J ) 2.0 Hz, HAr), 6.95 (d, 2H, J ) 7.5 Hz, HAr), 7.66 (d, 2H, J ) 7.5 Hz, HAr), 7.96 (s, 1H, dCH), 11.76 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 55.1, 55.4, 55.9, 90.0, 93.0, 104.6, 113.3 (2), 126.5, 129.0, 129.6 (2), 130.2, 140.5, 156.6, 158.7, 161.5, 161.9. MS: m/z 312 (M+ + 1). 5,8-Dimethoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinone (2f). According to the procedure described for 2a, the 2-quinolone 2f was prepared from 1f. IR (KBr): ν 1639, 1571 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.85 (s, 3H, CH3), 3.90 (s, 3H, CH3), 3.92 (s, 3H, CH3), 6.49 (d, 1H, J ) 8.7 Hz, HAr), 6.84 (d, 1H, J ) 8.7 Hz, HAr), 6.97 (d, 2H, J ) 8.8 Hz, HAr), 7.74 (d, 2H, J ) 8.8 Hz, HAr), 8.19 (s, 1H, dCH), 9.25 (br s, 1H, NH). 13C NMR (62.90 MHz, CDCl3): δ 55.2, 55.8, 56.2, 101.1, 109.7, 111.4, 113.6 (2), 128.7, 128.8, 130.0 (2), 131.4, 131.7, 139.4, 149.8, 159.5, 161.3. MS: m/z 312 (M+ + 1). 5,7-Dimethoxy-3-(4-benzyloxyphenyl)-1,2-dihydro-2quinolinone (2g). According to the procedure described for 2a, the 2-quinolone 2g was prepared from 1g. IR (KBr): ν 1629, 1608 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.81 (s, 3H, CH3), 3.90 (s, 3H, CH3), 5.15 (s, 2H, CH2), 6.37 (s, 1H, HAr), 6.45 (s, 1H, HAr), 7.04 (d, 2H, J ) 8.8 Hz, HAr), 7.317.49 (m, 5H, HAr), 7.69 (d, 2H, J ) 7.5 Hz, HAr), 7.97 (s, 1H, dCH), 11.76 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSOd6): δ 55.4, 55.9, 69.2, 90.0, 93.0, 104.6, 114.3 (2), 126.4, 127.6 (2), 127.8, 128.4 (2) 129.2, 129.6 (2), 130.2, 137.1, 140.5, 156.6, 157.7, 161.5, 161.9. MS: m/z 388 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1-methyl-1,2-dihydro-2-quinolinone (3). At 0 °C under argon, sodium hydride (93 mg, 3.86 mmol, 60% oil dispersion) was added portionwise to a solution of 2e (600 mg, 1.93 mmol) in anhydrous DMF (30 mL). The mixture was stirred for 15 min at 0 °C, and then, iodomethane (0.48 mL, 7.72 mmol) diluted in anhydrous DMF (5 mL) was added. The final solution was stirred at 90 °C and monitored by TLC (reaction time 18 h). The cooled mixture was partitioned between H2O (10 mL) and CH2Cl2 (10 mL). The organic layers were dried over MgSO4 and concentrated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/EtOAc 8:2) to give successively 3 (408 mg, 68%) and 3a (157 mg, 25%). IR (KBr):

ν 1635, 1596 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.73 (s, 3H, CH3), 3.83 (s, 3H, OCH3), 3.91 (s, 6H, OCH3), 6.30 (d, 1H, J ) 2.0 Hz, HAr), 6.37 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 7.5 Hz, HAr), 7.67 (d, 2H, J ) 7.5 Hz, HAr), 8.12 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 30.3, 55.3, 55.5, 55.8, 90.2, 92.6, 106.1, 113.5 (2), 127.0, 130.0, 130.1 (2), 130.2, 141.7, 157.6, 159.1, 162.2 (2). MS: m/z 326 (M+ + 1). 2,5,7-Trimethoxy-3-(4-methoxyphenyl)quinoline (3a). mp 106-107 °C (EtOAc/PE). IR (KBr): ν 1621, 1515 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.86 (s, 3H, OCH3), 3.94 (s, 6H, OCH3), 4.08 (s, 3H, OCH3), 6.40 (d, 1H, J ) 1.8 Hz, HAr), 6.85 (d, 1H, J ) 1.8 Hz, HAr), 6.97 (d, 2H, J ) 8.8 Hz, HAr), 7.57 (d, 2H, J ) 8.8 Hz, HAr), 8.26 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 53.5, 55.3, 55.5, 55.6, 95.9, 98.5, 112.8, 113.6 (2), 122.1, 129.6, 130.5 (2), 132.3, 147.9, 156.3, 158.9, 160.6, 161.3. MS: m/z 326 (M+ + 1). Anal. (C19H19NO4) C, H, N. Ethyl 2-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2dihydro-1-quinolinyl]acetate (4). According to the procedure described for 3a, the 2-quinolone 4 and compound 4a were prepared starting from 2e (1 equiv) and ethyl bromoacetate (2 equiv). Reaction time, 3 h; eluent chromatography CH2Cl2/ EtOAc 7:3; yield, 70%. IR (KBr): 1735, 1647, 1609 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.62 (t, 3H, J ) 7.1 Hz, CH3), 3.83 (s, 3H, OCH3), 3.87 (s, 3H, OCH3), 3.92 (s, 3H, OCH3), 4.22 (q, 2H, J ) 7.1 Hz, CH2), 5.10 (s, 2H, CH2), 6.14 (d, 1H, J ) 2.0 Hz, HAr), 6.30 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 7.5 Hz, HAr), 7.69 (d, 2H, J ) 7.5 Hz, HAr), 8.18 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 14.2, 44.8, 55.3, 55.5, 55.9, 61.6, 90.0, 92.8, 106.2, 113.5 (2), 126.6, 129.6, 130.1 (2), 131.0, 141.1, 157.8, 159.2, 161.9, 162.5, 168.4. MS: m/z 398 (M+ + 1). Ethyl 2-([5,7-Dimethoxy-3-(4-methoxyphenyl)-2-quinolinyl]oxy)acetate (4a). Yield, 25%; mp 95-96 °C (EtOAc/PE). IR (KBr): ν 1754, 1622, 1516 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.27 (t, 3H, J ) 7.1 Hz, CH3), 3,86 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 4.24 (q, 2H, J ) 7.1 Hz, CH2), 5.05 (s, 2H, CH2), 6.40 (d, 1H, J ) 2.0 Hz, HAr), 6.76 (d, 1H, J ) 2.0 Hz, HAr), 6.98 (d, 2H, J ) 9.0 Hz, HAr), 7.68 (d, 2H, J ) 9.0 Hz, HAr), 8.30 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 14.2, 55.3, 55.5, 55.7, 60.9, 62.7, 96.2, 98.6, 113.4, 113.7 (2), 121.7, 129.2, 130.6 (2), 132.8, 147.3, 156.3, 158.8, 159.1, 161.4, 169.5. MS: m/z 398 (M+ + 1). Anal. (C22H23NO6) C, H, N. N,N-Diethyl-2-[5,7-dimethoxy-3-(4-methoxyphenyl)-2oxo-1,2-dihydro-1-quinolinyl]acetamide (5). According to the procedure described for 3, the 2-quinolone 5 and compound 5a were prepared starting from 2e (1 equiv) and 2-chloro-N,Ndiethylacetamide (1.5 equiv). Reaction time, 3 h; eluent chromatography CH2Cl2/EtOAc 7:3; yield, 61%. IR (KBr): 1642, 1617, 1601 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.101.23 (m, 6H, CH3), 3.38-3.49 (m, 4H, CH2), 3.83 (s, 3H, OCH3), 3.86 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 5.17 (s, 2H, CH2), 6.28 (d, 1H, J ) 2.0 Hz, HAr), 6.34 (d, 1H, J ) 2.0 Hz, HAr), 6.93 (d, 2H, J ) 7.0 Hz, HAr), 7.66 (d, 2H, J ) 7.0 Hz, HAr), 8.17 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 13.0, 14.2, 40.9, 41.6, 45.3, 55.3, 55.5, 55.8, 90.8, 92.8, 106.3, 113.4 (2), 126.6, 129.9, 130.1 (2), 131.0, 141.7, 157.6, 159.0, 161.9, 162.3, 166.3. MS: m/z 425 (M+ + 1). N,N-Diethyl-2-([5,7-dimethoxy-3-(4-methoxyphenyl)-2quinolinyl]oxy)acetamide (5a). Yield, 30%; mp 146-147 °C (EtOAc/PE). IR (KBr): ν 1654, 1624, 1517 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.14 (t, 3H, J ) 7.5 Hz, CH3), 1.28 (t, 3H, J ) 7.5 Hz, CH3), 3.36-3.47 (m, 4H, CH2), 3.85 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 5.17 (s, 2H, CH2), 6.38 (d, 1H, J ) 2.0 Hz, HAr), 6.73 (d, 1H, J ) 2.0 Hz, HAr), 6.97 (d, 2H, J ) 9.0 Hz, HAr), 7.75 (d, 2H, J ) 9.0 Hz, HAr), 8.28 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 12.9, 14.2, 40.2, 55.2, 55.4, 55.6, 63.0, 95.8, 98.4, 113.3, 113.5 (2), 121.9, 129.2, 130.6 (2), 132.5, 147.3, 156.2, 158.9, 161.1, 167.3. MS: m/z 425 (M+ + 1). Anal. (C24H28N2O5), C, H, N. 1-[2-(Dimethylamino)ethyl]-5,7-dimethoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinone (6). According to the procedure described for 3, the 2-quinolone 6 and compound 6a were prepared starting from 2e (1 equiv) and 2-(dimethylamino)ethyl chloride (3 equiv). Reaction time, 3 h; eluent

3-Aryl-2-Quinolone Derivatives

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

2551

chromatography Et2O/EtOAc 3:7 and then CH2Cl2/MeOH 9:1; yield, 67%. IR (KBr): ν 1645, 1617 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.39 (s, 6H, CH3), 2.65 (t, 2H, J ) 7.8 Hz, CH2), 3.82 (s, 3H, OCH3), 3.90 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 4.44 (t, 2H, J ) 7.8 Hz, CH2), 6.28 (d, 1H, J ) 2.0 Hz, HAr), 6.49 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 8.8 Hz, HAr), 7.67 (d, 2H, J ) 8.8 Hz, HAr), 8.12 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 41.6, 45.8 (2), 55.3, 55.6, 55.8 (2), 90.1, 92.7, 106.4, 113.5 (2), 126.9, 129.8, 130.1 (2), 130.4, 141.1, 157.7, 159.1, 161.9, 162.4. MS: m/z 383 (M+ + 1). N,N-Dimethyl-2-[5,7-dimethoxy-3-(4-methoxyphenyl)2-quinolyl]oxy-1-ethanamine (6a). Yield, 30%; mp 49-50 °C (Et2O). IR (KBr): ν 1621, 1584 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.31 (s, 6H, NCH3), 2.77 (t, 2H, J ) 6.0 Hz, CH2), 3.85 (s, 3H, OCH3), 3.93 (s, 6H, OCH3), 4.62 (t, 2H, J ) 6.0 Hz, CH2), 6.39 (d, 1H, J ) 2.0 Hz, HAr), 6.81 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 8.8 Hz, HAr), 7.58 (d, 2H, J ) 8.8 Hz, HAr), 8.25 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 45.7 (2), 55.3, 55.5 (2), 57.8, 63.8, 95.9, 98.5, 112.9, 113.4 (2), 122.0, 129.5, 130.6 (2), 132.3, 147.8, 156.3, 158.9, 160.0, 161.3. MS: m/z 383 (M+ + 1). Anal. (C22H26N2O4) C, H, N. 1-[3-(Dimethylamino)propyl]-5,7-dimethoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinone (7). According to the procedure described for 3, the 2-quinolone 7 and compound 7a were prepared starting from 2e (1 equiv) and 3-(dimethylamino)propyl chloride (2.2 equiv). Reaction time, 3 h; eluent chromatography Et2O/MeOH 8:2 and then CH2Cl2/MeOH 9:1; yield, 70%. IR (KBr): ν 1635, 1617, 1598 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.91-2.04 (m, 2H, CH2), 2.28 (s, 6H, NCH3), 2.46 (t, 2H, J ) 7.2 Hz, CH2), 3.83 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 4.36 (t, 2H, J ) 7.2 Hz, CH2), 6.29 (d, 1H, J ) 2.0 Hz, HAr), 6.54 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 8.8 Hz, HAr), 7.68 (d, 2H, J ) 8.8 Hz, HAr), 8.13 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 25.5, 41.8, 45.6 (2), 55.4, 55.5, 55.8, 57.1, 90.3, 92.6, 106.4, 113.5 (2), 126.9, 130.0, 130.1 (2), 130.3, 141.1, 157.6, 159.1, 162.0, 162. MS: m/z 397 (M+ + 1). N,N-Dimethyl-2-[5,7-dimethoxy-3-(4-methoxyphenyl)2-quinolyl]oxy-1-propanamine (7a). Yield, 25%; mp 5455 °C (Et2O). IR (KBr): ν 1621, 1584 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.96-2.07 (m, 2H, CH2), 2.26 (s, 6H, NCH3), 2.47 (t, 2H, J ) 6.5 Hz, CH2), 3.84 (s, 3H, OCH3), 3.92 (s, 6H, OCH3), 4.53 (t, 2H, J ) 6.5 Hz, CH2), 6.39 (d, 1H, J ) 2.2 Hz, HAr), 6.82 (d, 1H, J ) 2.2 Hz, HAr), 6.96 (d, 2H, J ) 8.8 Hz, HAr), 7.57 (d, 2H, J ) 8.8 Hz, HAr), 8.26 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 27.0, 45.4 (2), 55.2, 55.5 (2), 56.6, 64.2, 95.8, 98.5, 112.7, 113.4 (2), 121.9, 129.6, 130.5 (2), 132.1, 147.9, 156.2, 158.8, 160.2, 161.2. MS: m/z 397 (M+ + 1). Anal. (C23H28N2O4) C, H, N. [5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2-dihydro1-quinolinyl]acetonitrile (8). According to the procedure described for 3, the 2-quinolone 8 and compound 8a were prepared starting from 2e (1 equiv) and bromoacetonitrile (2 equiv). Reaction time, 3 h; eluent chromatography CH2Cl2/ EtOAc 7:3; yield, 61%. IR (KBr): ν 2216, 1660, 1607 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.84 (s, 3H, OCH3), 3.94 (s, 3H, OCH3), 3.95 (s, 3H, OCH3), 5.29 (s, 2H, CH2), 6.36 (s, 2H, HAr), 6.95 (d, 2H, J ) 8.8 Hz, HAr), 7.65 (d, 2H, J ) 8.8 Hz, HAr), 8.17 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 30.4, 55.3, 55.8, 56.0, 90.0, 93.5, 106.2, 113.6 (2), 114.8, 126.3, 129.0, 130.0 (2), 131.7, 139.8, 158.1, 159.4, 161.1, 163.0. MS: m/z 351 (M+ + 1). 2-([5,7-Dimethoxy-3-(4-methoxyphenyl)-2-quinolinyl]oxy)acetonitrile (8a). Yield, 30%; mp 149-150 °C (Et2O). IR (KBr): ν 1623, 1586 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.87 (s, 3H, OCH3), 3.95 (s, 6H, OCH3), 5.17 (s, 2H, CH2), 6.45 (d, 1H, J ) 2.0 Hz, HAr), 6.87 (d, 1H, J ) 2.0 Hz, HAr), 7.99 (d, 2H, J ) 9.0 Hz, HAr), 7.54 (d, 2H, J ) 9.0 Hz, HAr), 8.34 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 50.1, 55.3, 55.6, 55.7, 96.9, 98.6, 113.8 (2), 113.9, 116.1, 121.3, 128.4, 130.5 (2), 133.5, 147.1, 156.3, 157.2, 129.3, 161.8. MS: m/z 351 (M+ + 1). Anal. (C20H18N2O4) C, H, N. 3-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2-dihydro-1-quinolinyl]butanenitrile (9). According to the pro-

cedure described for 3, the 2-quinolone 9 and compound 9a were prepared starting from 2e (1 equiv) and 4-chlorobutyronitrile (2 equiv). Reaction time, 3 h; eluent CH2Cl2/EtOAc 9:1; yield, 33%. IR (KBr): ν 2247, 1639, 1609, 1597 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.10-2.21 (m, 2H, CH2), 2.52 (t, 2H, J ) 7.2 Hz, CH2), 3.83 (s, 3H, OCH3), 3.92 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 4.43 (t, 2H, J ) 7.2 Hz, CH2), 6.31 (d, 1H, J ) 2.2 Hz, HAr), 6.42 (d, 1H, J ) 2.2 Hz, HAr), 6.94 (d, 2H, J ) 8.8 Hz, HAr), 7.66 (d, 2H, J ) 8.8 Hz, HAr), 8.15 (s, 1H, d CH). 13C NMR (62.90 MHz, CDCl3): δ 15.3, 23.6, 41.8, 55.4, 55.9, 56.0, 89.6, 93.1, 106.4, 113.6 (2), 119.5, 126.7, 129.6, 130.1 (2), 130.8, 140.7, 157.9, 159.3, 161.2, 162.8. MS: m/z 379 (M+ + 1). 2-([5,7-Dimethoxy-3-(4-methoxyphenyl)-2-quinolinyl]oxy)butanenitrile (9a). Yield, 33%; mp 89-90 °C (EtOAc). IR (KBr): ν 2247, 1624, 1607 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.12-2.22 (m, 2H, CH2), 2.50 (t, 2H, J ) 7.5 Hz, CH2), 3.87 (s, 3H, OCH3), 3.94 (s, 6H, OCH3), 4.61 (t, 2H, J ) 7.5 Hz, CH2), 6.40 (d, 1H, J ) 2.2 Hz, HAr), 6.81 (d, 1H, J ) 2.2 Hz, HAr), 6.97 (d, 2H, J ) 8.8 Hz, HAr), 7.53 (d, 2H, J ) 8.8 Hz, HAr), 8.27 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 14.6, 25.4, 55.4, 55.7, 55.8, 63.6, 96.2, 98.6, 113.1, 113.7 (2), 119.5, 122.0, 129.5, 130.5 (2), 132.8, 147.9, 156.4, 159.1, 159.7, 161.6. MS: m/z 379 (M+ + 1). Anal. (C22H22N2O4) C, H, N. Benzyl 2-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo1,2-dihydro-1-quinolinyl]acetate (10). According to the procedure described for 3, the 2-quinolone 10 and compound 10a were prepared starting from 2e (1 equiv) and benzyl bromoacetate (2 equiv). Reaction time, 2 h; eluent chromatography CH2Cl2; yield, 72%. IR (KBr): 1748, 1642, 1617 cm-1. 1H NMR (250 MHz, CDCl ): δ 3.68 (s, 3H, OCH ), 3.84 (s, 3H, 3 3 OCH3), 3.92 (s, 3H, OCH3), 5.16 (s, 2H, CH2), 5.21 (s, 2H, CH2), 6.03 (d, 1H, J ) 2.0 Hz, HAr), 6.27 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 7.5 Hz, HAr), 7.25-7.32 (m, 5H, HAr), 7.68 (d, 2H, J ) 7.5 Hz, HAr), 8.17 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 44.7, 55.3, 55.4, 55.8, 67.1, 89.8, 93.0, 106.2, 113.5 (2), 126.5, 128.3 (2), 128.4, 128.5 (2), 129.5, 130.1 (2), 131.1, 135.3, 141.0, 157.8, 159.2, 161.8, 162.4, 168.3. MS: m/z 460 (M+ + 1). Benzyl 2-([5,7-Dimethoxy-3-(4-methoxyphenyl)-2-quinolinyl]oxy)acetate (10a). Yield, 20%; mp 114-115 °C (Et2O). IR (KBr): ν 1761, 1621, 1517 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.87 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.95 (s, 3H, OCH3), 5.17 (s, 2H, CH2), 5.27 (s, 2H, CH2), 6.44 (d, 1H, J ) 2.0 Hz, HAr), 6.76 (d, 1H, J ) 2.0 Hz, HAr), 7.00 (d, 2H, J ) 8.0 Hz, HAr), 7.26-7.38 (m, 5H, HAr), 7.71 (d, 2H, J ) 8.0 Hz, HAr), 8.36 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 55.2, 55.4, 55.5, 62.6, 66.4, 96.2, 98.5, 113.4, 113.6 (2), 121.6, 128.0 (2), 128.4 (3), 129.0, 130.5 (2), 132.7, 135.6, 147.2, 156.1, 158.6, 159.0, 161.3, 169.3. MS: m/z 460 (M+ + 1). Anal. (C27H25NO6) C, H, N. 2-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2-dihydro-1-quinolinyl]acetic Acid (11). A solution of 10 (1.0 g, 2.2 mmol) in dry 1,4-dioxane (30 mL) was stirred under 40 psi of hydrogen for 4 h at room temperature in the presence of 10% Pd-C (100 mg) and then filtered. The filtrate was evaporated to dryness leaving a solid that was washed with Et2O to give 11 (764 mg, 95%). IR (KBr): ν 1732, 1614, 1583 cm-1. 1H NMR (250 MHz, DMSO-d6 + D2O): δ 3.77 (s, 3H, OCH3), 3.85 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 5.02 (s, 2H, CH2), 6.46 (s, 1H, HAr), 6.47 (s, 1H, HAr), 6.93 (d, 2H, J ) 9.0 Hz, HAr), 7.62 (d, 2H, J ) 9.0 Hz, HAr), 8.03 (s, 1H, dCH). 13C NMR (62.90 MHz, DMSO-d6): δ 44.9, 55.5, 56.1, 56.6, 91.3, 93.3, 105.3, 113.8 (2), 125.7, 129.4, 130.1 (2), 130.4, 141.3, 157.6, 159.1, 161.3, 162.8, 170.1. MS: m/z 370 (M+ + 1). Methyl 3-[5,7-Dimethoxy-3-(4-hydroxyphenyl)-2-oxo1,2-dihydro-1-quinolinyl]propanoate (12). To a solution of 2e (1.00 g, 3.2 mmol) and methyl acrylate (2.88 mL, 32.0 mmol) in anhydrous DMF (10 mL) at 0 °C was added Triton B (0.06 mL, 0.33 mmol). The final solution was stirred for 18 h at room temperature, and then, the solvent was removed in vacuo. The crude residue was partitioned between H2O and EtOAc and extracted. The organic layer was washed with H2O, dried over MgSO4, and concentrated in vacuo. The crude residue was

2552

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

Joseph et al.

purified by column chromatography (eluent CH2Cl2/EtOAc 8:2) to give 12 (1.06 g, 83%). IR (KBr): ν 1725, 1638 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.78 (t, 2H, J ) 8.0 Hz, CH2), 3.68 (s, 3H, CH3), 3.80 (s, 3H, OCH3), 3.88 (s, 6H, OCH3), 4.57 (t, 2H, J ) 8.0 Hz, CH2), 6.26 (d, 1H, J ) 2.0 Hz, HAr), 6.46 (d, 1H, J ) 2.0 Hz, HAr), 6.92 (d, 2H, J ) 8.8 Hz, HAr), 7.66 (d, 2H, J ) 8.8 Hz, HAr), 8.11 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 31.9, 39.1, 51.8, 55.2, 55.5, 55.7, 89.8, 92.6, 106.2, 113.4 (2), 126.5, 129.5, 129.9 (2), 130.3, 140.5, 157.6, 159.0, 161.7, 162.4, 172.0. MS: m/z 398 (M+ + 1). 3-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2-dihydro-1-quinolinyl]propanenitrile (13). According to the procedure described for 12, the 2-quinolone 13 was prepared starting from 2e (1 equiv) and acrylonitrile (10 equiv). Reaction time, 2 h; eluent chromatography CH2Cl2/EtOAc 7:3; yield, 63%. IR (KBr): ν 2241, 1639 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.88 (t, 2H, J ) 7.1 Hz, CH2), 3.84 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 3.94 (s, 3H, OCH3), 4.59 (t, 2H, J ) 7.1 Hz, CH2), 6.33 (d, 1H, J ) 1.8 Hz, HAr), 6.45 (d, 1H, J ) 1.8 Hz, HAr), 6.95 (d, 2H, J ) 9.0 Hz, HAr), 7.66 (d, 2H, J ) 9.0 Hz, HAr), 8.17 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 15.9, 39.4, 55.3, 55.7, 55.9, 89.9, 93.0, 106.4, 113.6 (2), 117.7, 126.6, 129.2, 130.0 (2), 131.1, 140.5, 158.0, 159.3, 161.8, 162.7. MS: m/z 365 (M+ + 1). Benzyl 3-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo1,2-dihydro-1-quinolinyl]propanoate (14). According to the procedure described for 12, the 2-quinolone 14 was prepared starting from 2e (1 equiv) and benzyl acrylate (10 equiv). Reaction time, 18 h; eluent chromatography EtOAc/PE 4:6; yield, 88%. IR (KBr): ν 1731, 1635, 1600 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.86 (t, 2H, J ) 8.0 Hz, CH2), 3.84 (s, 3H, OCH3), 3.85 (s, 3H, OCH3), 3.92 (s, 3H, OCH3), 4.63 (t, 2H, J ) 8.0 Hz, CH2), 5.14 (s, 2H, CH2), 6.29 (d, 1H, J ) 2.0 Hz, HAr), 6.49 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 7.5 Hz, HAr), 7.30-7.35 (m, 5H, HAr), 7.68 (d, 2H, J ) 7.5 Hz, HAr), 8.13 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 32.2, 39.1, 55.2, 55.4, 55.7, 66.5, 89.7, 92.6, 106.2, 113.4 (2), 126.5, 128.1 (2), 128.2, 128.4 (2), 129.5, 129.9 (2), 130.4, 135.5, 140.5, 157.6, 159.0, 161.7, 162.4, 171.3. MS: m/z 474 (M+ + 1). 3-[5,7-Dimethoxy-3-(4-methoxyphenyl)-2-oxo-1,2-dihydro-1-quinolinyl]propanoic Acid (15). A solution of 14 (1.0 g, 2.1 mmol) in dry 1,4-dioxane (30 mL) was stirred under 40 psi of hydrogen for 48 h at room temperature in the presence of 10% Pd-C (100 mg) and then filtered. The filtrate was evaporated to dryness leaving a solid that was washed with Et2O to give 15 (780 mg, 97%). IR (KBr): ν 1724, 1637, 1612, 1604 cm-1. 1H NMR (250 MHz, DMSO-d6 + D2O): δ 2.60 (t, 2H, J ) 7.5 Hz, CH2), 3.78 (s, 3H, OCH3), 3.90 (s, 3H, OCH3), 3.92 (s, 3H, OCH3), 4.48 (t, 2H, J ) 7.5 Hz, CH2), 6.48 (d, 1H, J ) 1.8 Hz, HAr), 6.62 (d, 1H, J ) 1.8 Hz, HAr), 6.95 (d, 2H, J ) 8.8 Hz, HAr), 7.62 (d, 2H, J ) 8.8 Hz, HAr), 8.00 (s, 1H, d CH). 13C NMR (62.90 MHz, DMSO-d6): δ 32.0, 38.9, 55.1, 55.7, 56.1, 90.6, 93.0, 105.0, 113.3 (2), 125.6, 129.3, 129.5, 129.8 (2), 140.5, 157.2, 158.7, 160.6, 162.4, 172.5. MS: m/z 384 (M+ + 1). 1-[2-(1H-1,2,3,4-Tetrazol-5-yl)ethyl]-5,7-dimethoxy-3-(4methoxyphenyl)-1,2-dihydro-2-quinolinone (16). A solution of 13 (350 mg, 0.9 mmol) and tributyltin azide (0,42 mL, 1.53 mmol) in anhydrous toluene (20 mL) was stirred at 105 °C for 65 h. After it was cooled, the solvent was removed in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/MeOH 9:1) to give 16 (333 mg, 85%). IR (KBr): ν 1618, 1594 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.33 (t, 2H, J ) 6.0 Hz, CH2), 3.79 (s, 3H, OCH3), 3.89 (s, 3H, OCH3), 3.93 (s, 3H, OCH3), 4.67 (t, 2H, J ) 6.0 Hz, CH2), 6.48 (s, 1H, HAr), 6.52 (s, 1H, HAr), 6.96 (d, 2H, J ) 9.0 Hz, HAr), 7.59 (d, 2H, J ) 9.0 Hz, HAr), 8.01 (s, 1H, dCH). 13C NMR (62.90 MHz, DMSO-d6): δ 21.4, 40.8, 55.1, 55.6, 56.1, 90.4, 93.0, 105.0, 113.3 (2), 125.5, 129.3, 129.6, 129.7 (2), 140.4, 153.7, 157.2, 158.7, 160.7, 162.4. MS: m/z 408 (M+ + 1). N,N-Diethyl-3-[5,7-dimethoxy-3-(4-methoxyphenyl)-2oxo-1,2-dihydro-1-quinolinyl]propanamide (17). To a solution of 15 (1.0 g, 2.6 mmol) in anhydrous DMF (25 mL) were added hydroxybenzotriazole (353 mg, 2.6 mmol) and dicyclo-

hexylcarbodiimide (540 mg, 2.6 mmol) at 0 °C. After the mixture was stirred at 0 °C for 10 min, N,N-diethylamine (0.26 mL, 2.6 mmol) was added and the final mixture was stirred at 0 °C for 2 h and at room temperature for 24 h. The white precipitate of dicyclohexylurea was removed by filtration. The solvent was evaporated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/EtOAc 7:3) to afford 17 (680 mg, 60%). IR (KBr): ν 1636, 1617 cm-1. 1H NMR (250 MHz, CDCl3): δ 1.11 (t, 3H, J ) 7.0 Hz, CH3), 1.13 (t, 3H, J ) 7.0 Hz, CH3), 2.78 (t, 2H, J ) 8.0 Hz, CH2), 3.30 (q, 2H, J ) 7.0 Hz, CH2), 3.39 (q, 2H, J ) 7.0 Hz, CH2), 3.84 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.94 (s, 3H, OCH3), 4.65 (t, 2H, J ) 8.0 Hz, CH2), 6.29 (d, 1H, J ) 2.0 Hz, HAr), 6.73 (d, 1H, J ) 2.0 Hz, HAr), 6.94 (d, 2H, J ) 7.5 Hz, HAr), 7.67 (d, 2H, J ) 7.5 Hz, HAr), 8.15 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 13.1, 14.4, 31.0, 40.1, 40.5, 42.2, 55.3, 55.8 (2), 89.9, 93.1, 106.3, 113.5 (2), 126.6, 129.7, 130.0 (2), 130.5, 140.9, 157.6, 159.1, 162.1, 162.6, 169.9. MS: m/z 439 (M+ + 1). 8,10-Dimethoxy-6-(4-methoxyphenyl)-2,3-dihydro1H,5H-pyrido[3,2,1-ij]quinoline-1,5-dione (18). In a dry round flask, P2O5 (63 mg) and PPA (500 mg) were stirred at 110 °C until complete dissolution of P2O5. Acid 15 (100 mg, 0.26 mmol) was added, and the final mixture was stirred at 110 °C for 45 min. After it was cooled, the mixture was poured into ice and neutralized by 2 N NaOH. CH2Cl2 (200 mL) was added, and the mixture was stirred overnight. After extraction, the aqueous layer was extracted with CH2Cl2 twice (2 × 50 mL). The organic phase was dried over MgSO4 and evaporated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/MeOH 98:2) to give 18 (62 mg, 65%). IR (KBr): ν 1678, 1649, 1634 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.83 (t, 2H, J ) 7.0 Hz, CH2), 3.85 (s, 3H, OCH3), 4.04 (s, 6H, OCH3), 4.54 (d, 2H, J ) 7.0 Hz, CH2), 6.32 (s, 1H, HAr), 6.96 (d, 2H, J ) 7.5 Hz, HAr), 7.68 (d, 2H, J ) 7.5 Hz, HAr), 8.12 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 37.6, 40.3, 55.3, 56.1, 56.5, 88.9, 103.5, 104.8, 113.6 (2), 127.6, 129.0, 129.8, 130.0 (2), 142.9, 159.4, 161.6, 161.7, 163.7, 190.3. MS: m/z 366 (M+ + 1). 5,7-Dihydroxy-3-(4-hydroxyphenyl)-1,2-dihydro-2-quinolinone (19).21 To a solution of 2e (1.0 g, 3.2 mmol) in AcOH (15 mL) was dropwise added 48% HBr in H2O (5 mL). The final solution was stirred at reflux for 3 days. The cooled mixture was diluted with H2O, neutralized with 10% aqueous NaOH (pH 6-7), and extracted with CH2Cl2 twice. The organic layer was dried over MgSO4 and then evaporated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/MeOH 9:1) to give 19 (320 mg, 37%). 1H NMR (250 MHz, DMSO-d6): δ 6.11 (d, 1H, J ) 2.0 Hz, HAr), 6.18 (d, 1H, J ) 2.0 Hz, HAr), 6.76 (d, 2H, J ) 8.6 Hz, HAr), 7.52 (d, 2H, J ) 8.6 Hz, HAr), 7.89 (s, 1H, dCH), 9.42 (s, 1H, OH), 9.84 (s, 1H, OH), 10.21 (s, 1H, OH), 11.46 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 91.3, 96.4, 103.5, 114.7 (2), 125.1, 127.8, 129.4 (2), 130.4, 140.7, 155.3, 156.6, 160.1, 161.8. MS: m/z 270 (M+ + 1). 5,7-Dihydroxy-3-(4-hydroxyphenyl)-1-methyl-1,2-dihydro-2-quinolinone (20). According to the procedure described for 19, the 2-quinolone 20 was prepared starting from 3 (eluent CH2Cl2/MeOH 9:1); yield: 35%. 1H NMR (250 MHz, DMSOd6): δ 3,53 (s, 3H, NCH3), 6.25 (s, 2H, HAr), 6.76 (d, 2H, J ) 8.8 Hz, HAr), 7.47 (d, 2H, J ) 8.8 Hz, HAr), 7.92 (s, 1H, dCH), 9.42 (s, 1H, OH), 10.00 (s, 1H, OH), 10.35 (s, 1H, OH). 13C NMR (62.90 MHz, DMSO-d6): δ 29.7, 91.8, 96.5, 103.5, 114.7 (2), 124.3, 128.4, 129.7 (3), 141.7, 155.9, 156.7, 160.6, 161.1. MS: m/z 284 (M+ + 1). 5,7-Dimethoxy-3-(4-hydroxyphenyl)-1,2-dihydro-2-quinolinone (21). To a solution of 2e (530 mg, 1.70 mmol) in AcOH (15 mL) was dropwise added 48% HBr in H2O (2.7 mL). The final solution was stirred at reflux for 5 h. The cooled mixture was diluted with H2O, neutralized with 10% aqueous NaOH (pH 6-7), and extracted with CH2Cl2 twice. The organic layer was dried over MgSO4 and then evaporated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/MeOH 9:1) to give 21 (175 mg, 35%). IR (KBr): ν 1628, 1604, 1558 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.80 (s,

3-Aryl-2-Quinolone Derivatives

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

2553

3H, OCH3), 3.89 (s, 3H, OCH3), 6.35 (d, 1H, J ) 2.5 Hz, HAr), 6.43 (d, 1H, J ) 2.5 Hz, HAr), 6.78 (d, 2H, J ) 7.5 Hz, HAr), 7.55 (d, 2H, J ) 7.5 Hz, HAr), 7.92 (s, 1H, dCH), 9.48 (s, 1H, OH), 11.72 (br s, 1H, NH). 13C NMR (62.90 MHz, DMSO-d6): δ 55.4, 55.9, 90.0, 93.0, 104.7, 114.8 (2), 126.9, 127.4, 129.6, 129.8 (2), 140.4, 156.6, 156.9, 161.6, 161.8. MS: m/z 298 (M+ + 1). 5,7-Dimethoxy-3-(4-hydroxyphenyl)-1-methyl-1,2-dihydro-2-quinolinone (22). According to the procedure described for 21, the 2-quinolone 22 was prepared starting from 3 (eluent CH2Cl2/EtOAc 7:3); yield, 56%. 1H NMR (250 MHz, DMSOd6): δ 3.63 (s, 3H, NCH3), 3.89 (s, 3H, OCH3), 3.90 (s, 3H, OCH3), 6.46 (d, 1H, J ) 1.9 Hz, HAr), 6.54 (d, 1H, J ) 1.9 Hz, HAr), 6.76 (d, 2H, J ) 8.5 Hz, HAr), 7.48 (d, 2H, J ) 8.5 Hz, HAr), 7.93 (s, 1H, dCH), 9.47 (s, 1H, OH). 13C NMR (62.90 MHz, DMSO-d6): δ 30.5, 56.1, 56.5, 91.3, 93.4, 105.3, 115.2 (2), 126.5, 128.4, 129.2, 130.2 (2), 141.7, 157.4 (2), 161.4, 162.5. MS: m/z 312 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1,2-dihydro-2-quinolinethione (23). A mixture of 2e (100 mg, 0.32 mmol) and Lawesson’s reagent (260 mg, 0.64 mmol) in anhydrous toluene (15 mL) was stirred at reflux for 18 h. After it was cooled, the solvent was removed in vacuo and the residue was purified by silica gel chromatography (eluent CH2Cl2/EtOAc 9:1) to give 23 (81 mg, 77%). IR (KBr): ν 1636, 1610, 1524 cm-1. 1H NMR (250 MHz, DMSO-d6): δ 3.78 (s, 3H, OCH3), 3.83 (s, 3H, OCH3), 3.89 (s, 3H, OCH3), 6.49 (d, 1H, J ) 1.9 Hz, HAr), 6.79 (d, 1H, J ) 1.9 Hz, HAr), 6.92 (d, 2H, J ) 8.7 Hz, HAr), 7.49 (d, 2H, J ) 8.7 Hz, HAr), 7.78 (s, 1H, dCH), 13.50 (br s, 1H, NH). 13C NMR (62.90 MHz, CDCl ): δ 55.1, 55.6, 56.1, 90.2, 95.2, 3 108.9, 112.9 (2), 128.2, 130.7 (2), 132.0, 136.3, 140.9, 156.4, 158.5, 162.5, 180.5. MS: m/z 328 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1-methyl-1,2-dihydro-2-quinolinethione (24). According to the procedure described for 23, the 2-thioquinolone 24 was prepared starting from 3 (eluent EtOAc/PE 3:7); yield, 72%. IR (KBr): ν 1613, 1570, 1512 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.84 (s, 3H, OCH3), 3.91 (s, 3H, OCH3), 3.95 (s, 3H, OCH3), 4.39 (s, 3H, NCH3), 6.39 (d, 1H, J ) 2.0 Hz, HAr), 6.56 (d, 1H, J ) 2.0 Hz, HAr), 6.93 (d, 2H, J ) 7.5 Hz, HAr), 7.43 (d, 2H, J ) 7.5 Hz, HAr), 7.97 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 39.5, 55.2, 55.6, 55.9, 91.1, 94.6, 109.9, 113.1 (2), 126.6, 130.8 (2), 134.3, 138.6, 142.7, 157.6, 158.8, 162.7, 184.5. MS: m/z 342 (M+ + 1). 5,7-Acetyl-3-(4-acetylphenyl)-1,2-dihydro-2-quinolinone (25). A solution of 20 (200 mg, 0.71 mmol), anhydride acetic, and pyridine (8 mL, v/v) was stirred at room temperature for 18 h. H2O (10 mL) was added to the mixture and then extracted with CH2Cl2 (2 × 10 mL). The organic layer was dried over MgSO4 and then evaporated in vacuo. The crude residue was purified by column chromatography (eluent CH2Cl2/EtOAc 9:1) to give 25 (220 mg, 76%). IR (KBr): ν 1769, 1748, 1638, 1598 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.31 (s, 3H, CH3), 2.33 (s, 3H, CH3), 2.40 (s, 3H, CH3), 3.72 (s, 3H, NCH3), 6.92 (d, 1H, J ) 2.0 Hz, HAr), 7.03 (d, 1H, J ) 2.0 Hz, HAr), 7.14 (d, 2H, J ) 8.5 Hz, HAr), 7.67 (d, 2H, J ) 8.5 Hz, HAr), 7.78 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 20.8, 21.0, 21.2, 30.5, 105.2, 110.2, 111.8, 121.4 (2), 129.7, 130.3 (2), 131.5, 134.2, 141.0, 148.8, 150.7, 151.9, 161.2, 168.6, 168.8, 169.6. MS: m/z 410 (M+ + 1). 5,7-Dimethoxy-3-(4-acetylphenyl)-1,2-dihydro-2-quinolinone (26). According to the procedure described for 25, the 2-quinolone 26 was prepared starting from 22. Yield, 73%; reaction time, 2 h; eluent CH2Cl2/EtOAc 9:1. IR (KBr): ν 1751, 1639, 1601 cm-1. 1H NMR (250 MHz, CDCl3): δ 2.31 (s, 3H, CH3), 3.72 (s, 3H, NCH3), 3.90 (s, 6H, OCH3), 6.28 (d, 1H, J ) 2.0 Hz, HAr), 6.34 (d, 1H, J ) 2.0 Hz, HAr), 7.13 (d, 2H, J ) 8.8 Hz, HAr), 7.75 (d, 2H, J ) 8.8 Hz, HAr), 8.15 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 21.3, 30.4, 55.7, 55.9, 90.3, 92.7, 106.0, 121.1 (2), 126.3, 130.1 (3), 131.4, 142.1, 150.1, 157.8, 162.0, 162.7, 169.6. MS: m/z 354 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1-methyl-2-(methylsulfanyl)quinolinium Iodide (27). To a solution of 24 (682 mg, 2.0 mmol) in anhydrous THF (30 mL) was added io-

domethane (3.5 mL, 56 mmol) diluted in anhydrous THF (5 mL) at room temperature. The final solution was stirred overnight at room temperature. The red precipitate obtained was collected and washed with the same solvent to provide 27 (761 mg, 79%); mp 156-157 °C (THF washing). 1H NMR (250 MHz, CDCl3): δ 2.44 (s, 3H, SCH3), 3.88 (s, 3H, OCH3), 4.02 (s, 3H, OCH3), 4.28 (s, 3H, OCH3), 5.03 (s, 3H, NCH3), 6.70 (d, 1H, J ) 1.5 Hz, HAr), 7.03 (d, 2H, J ) 8.5 Hz, HAr), 7.37 (br s, 1H, HAr), 7.45 (d, 2H, J ) 8.5 Hz, HAr), 8.71 (s, 1H, dCH). 13C NMR (62.90 MHz, CDCl3): δ 20.8, 46.5, 55.4, 56.7, 58.5, 92.9, 101.0, 114.4 (2), 117.6, 125.8, 128.9, 130.7 (2), 135.9, 138.8, 144.2, 157.4, 160.3, 167.7. 5,7-Dimethoxy-3-(4-methoxyphenyl)-1-methyl-1,2-dihydro-2-quinolinone-2-phenylhydrazone (28). In a sealed tube, a solution of 27 (200 mg, 0.4 mmol) and phenylhydrazine (0.28 mL, 2.8 mmol) in anhydrous EtOH (5 mL) was heated at 90 °C overnight. After it was cooled, the solvent was removed in vacuo. The residue was dissolved in CH2Cl2 (10 mL), and the organic solution was washed with H2O and then with saturated NaHCO3 solution twice. The organic phase was dried over MgSO4 and evaporated in vacuo. The crude residue was crystallized from MeOH to afford 28 (102 mg, 60%). IR (KBr): ν 3340, 1602, 1599 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.58 (s, 3H, NCH3), 3.85 (s, 3H, OCH3), 3.86 (s, 3H, OCH3), 3.88 (s, 3H, OCH3), 6.04 (s, 1H, HAr), 6.14 (s, 1H, HAr), 6.48 (br s, 1H, NH), 6.56-6.67 (m, 3H, HAr), 6.94 (d, 2H, J ) 8.8 Hz, HAr), 7.11 (t, 2H, J ) 7.7 Hz, HAr), 7.28 (s, 1H, dCH), 7.287.34 (m, 2H, HAr). 13C NMR (62.90 MHz, CDCl3): δ 33.3, 55.4 (2), 55.6, 89.6 (2), 111.6, 113.8 (2), 117.8, 125.9, 128.3, 128.9 (3), 129.5 (2), 130.5, 146.2, 157.2, 159.2 (2), 162.3. MS: m/z 416 (M+ + 1). 5,7-Dimethoxy-3-(4-methoxyphenyl)-1-methyl-1,2-dihydro-2-quinolinone-2-(2-pyridinyl)hydrazone (29). Compound 29 was obtained according to the procedure described below but substituting the phenylhydrazine by 2-hydrazinopyridine; yield, 58%. IR (KBr): 3353, 1628, 1593 cm-1. 1H NMR (250 MHz, CDCl3): δ 3.61 (s, 3H, NCH3), 3.87 (s, 6H, OCH3), 3.90 (s, 3H, OCH3), 6.07 (s, 1H, HAr), 6.17 (s, 1H, HAr), 6.516.55 (m, 1H, HPyr), 6.94-7.01 (m, 3H, HAr + HPyr), 7.14 (br s, 1H, NH), 7.32-7.36 (m, 3H, dCH + HAr) 7.48 (t, 1H, J ) 7.3 Hz, HPyr), 7.91 (d, 1H, J ) 4.3 Hz, HPyr). 13C NMR (62.90 MHz, CDCl3): δ 33.2, 55.3, 55.4, 55.6, 89.7 (2), 104.7, 105.7, 113.4, 114.1 (2), 122.9, 129.0, 129.2 (2), 130.1, 137.5, 140.7, 143.7, 147.8, 157.3, 157.4, 159.2, 162.4. MS: m/z 417 (M+ + 1). Pharmacology. In Vitro MTT Colorimetric Assay. Twelve human tumor cell lines were obtained from the American Type Culture Collection (ATCC, Manassas, VA). These included three glioblastomas (A-172, U-373 MG, and U-87 MG), two colon (HCT-15 and LoVo), two nonsmall cell lung (A549 and A-427), two bladder (J82 and T24), one prostate (PC-3), and two breast (T-47D and MCF-7) cancer models. The ATCC numbers of these cell lines are CRL1620 (A-172), HTB 14 (U-87 MG), HTB 17 (U-373 MG), CCL225 (HCT-15), CCL229 (LoVo), CCL 185 (A549), HBT 53 (A-427), HTB1 (J82), HTB4 (T24), HTB133 (T-47D), HTB22 (MCF-7), and CRL1435 (PC-3). The cells were cultured at 37 °C in sealed (airtight) Falcon plastic dishes (Nunc, Gibco, Belgium) containing Eagle’s minimal essential medium (MEM, Gibco) supplemented with 5% fetal calf serum (FCS). All of the media were supplemented with a mixture of 0.6 mg/mL glutamine (Gibco), 200 IU/mL penicillin (Gibco), 200 IU/mL streptomycin (Gibco), and 0.1 mg/mL gentamycin (Gibco). The FCS was heat-inactivated for 1 h at 56 °C. The twelve cell lines were incubated for 24 h in 96 microwell plates (at a concentration of 40 000 cells/mL culture medium) to ensure adequate plating prior to cell growth determination, which was carried out by means of the colorimetric MTT assay as detailed previously.23 Six concentrations ranging from 10-5 to 10-9 M were assayed for each of the 25 drugs under study. In Vitro Cell Migration Assay. The cell motility level of the MCF-7 human breast cancer cells was quantitatively determined by means of a device detailed elsewhere.24 Briefly, this device consists of an inverted-phase contrast microscope equipped with a black and white CCD camera, an incubator

2554

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

maintaining the temperature at 37 °C, a computer containing a frame grabber, and an image-processing software that analyzes digitized frames. The software that we set up thus enabled each living cell in the culture under study to be isolated automatically on the basis of specific morphological characteristics that in turn enables cells to be identified against their background (the plastic Falcon dishes). Once the segmentation procedure is complete, each cell is transformed into its center of gravity. An image is digitized every 4 min by the computer, and the video system is thus able to track the trajectory of each cell by analyzing the movement of its center of gravity. From these trajectories, the maximum relative distance to the origin (the MRDO quantitative variable) of the cell was calculated for each cell under analysis. This variable thus describes the greatest linear distance found between the original and the subsequent positions of the cell divided by the observation time.24 All of the experiments were performed over 48 h at a cell concentration of 10 000 MCF-7 cells/mL MEM, with one image recorded every 4 min. The MCF-7 cells were grown in a standard MEM medium (see above for the MTT colorimetric assay) in control or in culture media containing either compound 13 or 16 at 10-6, 10-7, or 10-8 M concentration. Each experimental condition was carried out in triplicate. At the beginning of the experiment, the minimal number of cells in a given experimental condition was 34, and the maximal number at the end of the experiments was 402 cells. According to the analyses in triplicate, the trajectories of a minimum of 719 and a maximum of 1002 MCF-7 cells were analyzed in each experimental condition. In Vivo Determination of the MTD. We determined the MTD for each of 25 quinolone derivatives under study by defining the maximum dose of the drug that can be administered acutely (i.e., in one i.p. single dose) to healthy animals (B6D2F1, Iffa Credo), i.e., not grafted with tumors. The survival and weight of the animals were recorded for up to 28 days postinjection. Six different doses of each drug (5, 10, 20, 40, 80, and 160 mg/kg) were used for the determination of the MTD index, with each experimental group composed of three mice for this purpose. In Vivo Determination of Antitumor Activity. The hormone-sensitive MXT (MXT-HS) mammary cancer model was set up experimentally at Baylor College by the Clark group.37 We obtained the MXT-HS model in 1983 from Dr. D. Bogden (Mason Research Institute, Worcester, MA). The MXTHS strain died out in our laboratory after a number of years, and only the hormone-insensitive (MXT-HI) one continued.36 An experimental protocol was thus developed in our laboratory to differentiate hormone-insensitive MXT-HI tumor strains into hormone-sensitive MXT-HS ones.32 The MXT-HS tumors that we set up from the MXT-HI ones are maintained in our laboratory by monthly s.c. transplantations into 6 week old female B6D2F1 mice (Iffa Credo). Tumor size was measured weekly by means of a caliper and expressed as an area (mm2) by multiplying together the two largest perpendicular diameters. All of the animals were kept in plastic cages in a room with a controlled temperature (22 ( 1 °C), light exposure (from 6:00 am to 6:00 pm), and 40-70% relative humidity. Food (AO4, UAR, Villemoisson, France) and water were provided ad libitum. Statistical Analysis. The results are presented as the mean ( the standard error of the mean (SEM). The statistical comparisons of the data were carried out by means of the Fisher F (one way variance analysis for more than two groups) or the Student t (for two groups) tests after a check of the homogeneity of variance by means of the Levene test and of the normal distribution fitting of the data by means of the χ squared test of goodness-of-fit. When these parametric conditions were not satisfied, the nonparametric Kruskall-Wallis (for more than two groups) or the Mann-Whitney (for two groups) tests were carried out. All of the statistical analyses were carried out using Statistica (Statsoft, Tulsa, OK).

Joseph et al.

References (1) Fidler, I. J. Molecular biology of cancer: Invasion and metastasis. In Cancer: Principles and Practice of Oncology, 5th ed.; DeVita, V. T., Jr., Hellman, S., Rosenberg, S. A., Eds.; JB Lippincott Co.: Philadelphia, 1997; pp 135-152. (2) Folkman, J.; Shing, Y. Angiogenesis. Minireview. J. Biol. Chem. 1992, 267, 10931-10934. (3) Chambers, A. F.; Matrisian, L. M. Changing views of the role of matrix metalloproteinases in metastasis. J. Natl. Cancer Inst. 1997, 89, 1260-1270. (4) DeVita, V. T., Jr. Principles of cancer management: Chemotherapy. In Cancer: Principles and Practice of Oncology, 5th ed.; DeVita, V. T., Jr., Hellman, S., Rosenberg, S. A., Eds.; JB Lippincott Co.: Philadelphia, 1997; pp 333-348. (5) Kerr, J. F.; Wyllie, A. H.; Currie, A. R. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239-257. (6) Wyllie, A. H.; Kerr, J. F.; Currie, A. R. Cell death: the significance of apoptosis. Int. Rev. Cytol. 1980, 68, 251-306. (7) Hande, K. R. Clinical applications of anticancer drugs targeted to topoisomerase II. Biochim. Biophys. Acta 1998, 1400, 173184. (8) Malonne, H.; Atassi, G. DNA topoisomerase targeting drugs: mechanisms of action and perspectives. Anticancer Drugs 1997, 8, 811-822. (9) Folkman, J. Clinical applications of research on angiogenesis. New Engl. J. Med. 1995, 333, 1757-1763. (10) Bremers, A. J.; Kuppen, P. J.; Parmiani, G. Tumour immunotherapy: the adjuvant treatment of the 21st century? Eur. J. Surg. Oncol. 2000, 26, 418-424. (11) Curiel, D. T.; Gerritsen, W. R.; Krul, M. R. Progress in cancer gene therapy. Cancer Gene Ther. 2000, 7, 1197-1199. (12) Downing, K. H. Structural basis for the action of drugs that affect microtubule dynamics. Emerging Ther. Targets 2000, 4, 219237. (13) Hertog, M. G. L.; Hollman, P. C. H.; Katan, M. B.; Kromhout, D. Intake of potentially anticarcinogenic flavonoids and their determinants in adults in The Netherlands. Nutr. Cancer 1993, 20, 21-29. (14) So, F. V.; Guthrie, N.; Chambers, A. F.; Carroll, K. K. Inhibition of proliferation of estrogen receptor-positive MCF-7 human breast cancer cells by flavonoids in the presence and absence of excess estrogen. Cancer Lett. 1997, 112, 127-133. (15) Yoshida, M.; Yamamoto, N.; Nikaido, T. Quercetin arrests human leukemic T-cells in late G1-phase of the cell cycle. Cancer Res. 1992, 52, 6676-6681. (16) Alonso, M. A.; Blanco, M. M.; Avendan˜o, C.; Menen˜dez, J. C. Synthesis of 2,5,8(1H)-quinolinetrione derivatives through Vilsmeier-Haack formylation of 2,5-dimethoxyanilides. Heterocycles 1993, 36, 2315-2325. (17) Uray, G.; Niederreiter, K. S.; Belaj, F.; Fabian, W. M. F. Longwavelength-absorbing and -emitting carbostyrils with high fluorescence quantum yields. Helv. Chim. Acta 1999, 82, 14081417. (18) Yamagami, C.; Takao, N.; Tanaka, M.; Horisaka, K.; Asada, S.; Fujita, T. A quantitative structure-activity study of anticonvulsant phenylacetanilides. Chem. Pharm. Bull. 1984, 32, 50035009. (19) Taber, D. F.; Mack, J. F.; Rheingolf, A. L.; Geib, S. J. Enantioselective Robinson annulation: Synthesis of (+)-O-methyljoubertiamine. J. Org. Chem. 1989, 54, 3831-3836. (20) Imamoto, T.; Matsumoto, T.; Yokoyama, H.; Yokoyama, M.; Yamaguchy, K. Preparation and synthetic use of trimethylsilyl polyphosphate. A new stereoselective aldol-type reaction in the presence of trimethyl silyl polyphosphate. J. Org. Chem. 1984, 49, 1105-1110. (21) Croisy, M.; Huel, C.; Bisagni, E. Synthesis of 3-(4-methoxyphenyl)-5,7-dimethoxy-(1H)-quinolin-2 or 4-ones and derivatives. Heterocycles 1997, 45, 683-690. (22) Ferna´ndez, M.; de la Cuesta, E.; Avendan˜o, C. Synthesis of 5-methoxy-2(1H)-quinolinone. Heterocycles 1994, 38, 2615-2620. (23) Camby, I.; Salmon, I.; Danguy, A.; Pasteels, J. L.; Brotchi, J.; Martinez, J.; Kiss, R. Influence of gastrin on human astrocytic tumor cell proliferation. J. Natl. Cancer Inst. 1996, 88, 594600. (24) DeHauwer, C.; Camby, I.; Darro, F.; Decaestecker, C.; Gras, T.; Salmon, I.; Kiss, R.; VanHam, P. Dynamic characterization of glioblastoma cell motility. Biochem. Biophys. Res. Commun. 1997, 232, 267-272. (25) Belot, N.; Rorive, S.; Doyen, I.; Lefranc, F.; Bruyneel, E.; Dedecker, R.; Micik, S.; Brotchi, J.; Decaestecker, C.; Salmon, I.; Kiss, R.; Camby, I. Molecular characterization of cellsubstratum attachments in human glial tumors relates to prognostic features. GLIA 2001, 36, 375-390. (26) Frasca, F.; Vigneri, P.; Vella, V.; Vigneri, R.; Wang, J. Y. Tyrosine kinase inhibitor STI571 enhances thyroid cancer cell motile response to hepatocyte growth factor. Oncogene 2001, 20, 38453856.

3-Aryl-2-Quinolone Derivatives (27) Kermorgant, S.; Aparicio, T.; Dessirier, V.; Lewin, M. J.; Lehy, T. Hepatocyte growth factor induces colonic cancer cell invasiveness via enhanced motility and protease overproduction. Evidence for PI3 kinase and PKC involvement. Carcinogenesis 2001, 22, 1035-1042. (28) Huang, L.-J.; Hsieh, M.-C.; Teng, C.-M.; Lee, K.-H.; Kuo, S.-C. Synthesis and antiplatelet activity of phenyl quinolones. Bioorg. Med. Chem. 1998, 6, 1657-1662. (29) Boschelli, D. H.; Wu, Z.; Klutchko, S. R.; Showalter, H. D.; Hamby, J. M.; Lu, G. H.; Major, T. C.; Dahring, T. K.; Batley, B.; Panek, R. L.; Keiser, J.; Hartl, B. G.; Kraker, A. J.; Klohs, W. D.; Roberts, B. J.; Patmore, S.; Elliot, W. L.; Steinkampf, R.; Bradford, L. A.; Hallak, H.; Doherty, A. M. Synthesis and tyrosine kinase inhibitory activity of a series of 2-amino-8Hpyrido[2,3-d]pyrimidines: identification of potent, selective platelet-derived growth factor receptor tyrosine kinase inhibitors. J. Med. Chem. 1998, 41, 4365-4377. (30) Cheresh, D. A.; Eliceiri, B.; Paul, R. Angiogenis and vasculor permeability modulators and inhibitors. PCT Int. Appl. WO 0145751, 2001 (Chem. Abst. 135: 81944). (31) Kiss, R.; de Launoit, Y.; L’Hermite-Bale´riaux, M.; L’Hermite, M.; Paridaens, R.; Danguy, A.; Pasteels, J. L. Effect of prolactin and estradiol on cell proliferation in the uterus and the MXT mouse mammary neoplasm. J. Natl. Cancer Inst. 1987, 78, 993998.

Journal of Medicinal Chemistry, 2002, Vol. 45, No. 12

2555

(32) Kiss, R.; de Launoit, Y.; Danguy, A.; Paridaens, R.; Pasteels, J. L. Influence of pituitary grafts or prolactin administrations on the hormone sensitivity of ovarian hormone-independent mouse mammary MXT tumors. Cancer Res. 1989, 49, 2945-2951. (b) Watson, C. S.; Medina, D.; Clarck, J. H. Estrogen characterization in a transplantable mouse mammary tumor. Cancer Res. 1977, 37, 3344-3348. (33) Paridaens, R.; Leclercq, G.; Piccart, M.; Kiss, R.; Mattheiem, W.; Heuson, J. C. Comments on the treatment of breast cancer. In Hormones of Cancer: 90 Years after Beatson (Bulbrook, R. D., Ed.). Cancer Surveys 1986, 5, 447-461. (34) Briand, P. Hormone-dependent mammary tumors in mice and rats as a model for human breast cancer (Review). Anticancer Res. 1983, 3, 273-282. (35) Pihl, A. UICC Study group on chemosensitivity testing of human tumors. Problems-Applications-Future prospects. Int. J. Cancer 1986, 37, 1-5. (36) Danguy, A.; Kiss, R.; Leclercq, G.; Heuson, J. C.; Pasteels, J. L. Morphology of MXT mouse mammary tumors.Correlation with growth characteristics and hormone sensitivity. Eur. J. Cancer Clin. Oncol. 1986, 22, 69-76. (37) Watson, C. S.; Medina, D.; Clarck, J. H. Estrogen characterization in a transplantable mouse mammary tumor. Cancer Res. 1977, 37, 3344-3348.

JM010978M